Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Front Cell Infect Microbiol ; 14: 1375887, 2024.
Article in English | MEDLINE | ID: mdl-38505286

ABSTRACT

Salmonella enterica is a food-borne pathogen able to cause a wide spectrum of diseases ranging from mild gastroenteritis to systemic infections. During almost all stages of the infection process Salmonella is likely to be exposed to a wide variety of host-derived antimicrobial peptides (AMPs). AMPs are important components of the innate immune response which integrate within the bacterial membrane, thus forming pores which lead ultimately to bacterial killing. In contrast to other AMPs Bactericidal/Permeability-increasing Protein (BPI) displayed only weak bacteriostatic or bactericidal effects towards Salmonella enterica sv. Typhimurium (STM) cultures. Surprisingly, we found that sub-antimicrobial concentrations of BPI fold-containing (BPIF) superfamily members mediated adhesion of STM depending on pre-formed type 1 fimbriae. BPIF proteins directly bind to type 1 fimbriae through mannose-containing oligosaccharide modifications. Fimbriae decorated with BPIF proteins exhibit extended binding specificity, allowing for bacterial adhesion on a greater variety of abiotic and biotic surfaces likely promoting host colonization. Further, fimbriae significantly contributed to the resistance against BPI, probably through sequestration of the AMP before membrane interaction. In conclusion, functional subversion of innate immune proteins of the BPIF family through binding to fimbriae promotes Salmonella virulence by survival of host defense and promotion of host colonization.


Subject(s)
Salmonella enterica , Salmonella typhimurium , Fimbriae, Bacterial/metabolism , Bacterial Adhesion , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism
2.
PLoS Pathog ; 19(6): e1011468, 2023 06.
Article in English | MEDLINE | ID: mdl-37384799

ABSTRACT

Controlled human malaria infections (CHMI) are a valuable tool to study parasite gene expression in vivo under defined conditions. In previous studies, virulence gene expression was analyzed in samples from volunteers infected with the Plasmodium falciparum (Pf) NF54 isolate, which is of African origin. Here, we provide an in-depth investigation of parasite virulence gene expression in malaria-naïve European volunteers undergoing CHMI with the genetically distinct Pf 7G8 clone, originating in Brazil. Differential expression of var genes, encoding major virulence factors of Pf, PfEMP1s, was assessed in ex vivo parasite samples as well as in parasites from the in vitro cell bank culture that was used to generate the sporozoites (SPZ) for CHMI (Sanaria PfSPZ Challenge (7G8)). We report broad activation of mainly B-type subtelomeric located var genes at the onset of a 7G8 blood stage infection in naïve volunteers, mirroring the NF54 expression study and suggesting that the expression of virulence-associated genes is generally reset during transmission from the mosquito to the human host. However, in 7G8 parasites, we additionally detected a continuously expressed single C-type variant, Pf7G8_040025600, that was most highly expressed in both pre-mosquito cell bank and volunteer samples, suggesting that 7G8, unlike NF54, maintains expression of some previously expressed var variants during transmission. This suggests that in a new host, the parasite may preferentially express the variants that previously allowed successful infection and transmission. Trial registration: ClinicalTrials.gov - NCT02704533; 2018-004523-36.


Subject(s)
Culicidae , Malaria, Falciparum , Malaria , Parasites , Animals , Humans , Culicidae/genetics , Gene Expression , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Parasites/genetics , Plasmodium falciparum/genetics , Sporozoites , Virulence/genetics
3.
Infect Immun ; 82(7): 2763-71, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24733095

ABSTRACT

Intracellular bacterial pathogens frequently inhibit host cell apoptosis to ensure survival of their host, thereby allowing bacterial propagation. The obligate intracellular pathogen Coxiella burnetii displays antiapoptotic activity which depends on a functional type IV secretion system (T4SS). Accordingly, antiapoptotic T4SS effector proteins, like AnkG, have been identified. AnkG inhibits pathogen-induced apoptosis, possibly by binding to the host cell mitochondrial protein p32 (gC1qR). However, the molecular mechanism of AnkG activity remains unknown. Here, we demonstrate that ectopically expressed AnkG associates with mitochondria and traffics into the nucleus after apoptosis induction, although AnkG lacks a predicted nuclear localization signal. We identified the p32 interaction region in AnkG and constructed an AnkG mutant (AnkGR(22/23S)) unable to bind to p32. By using this mutant, we found that intracellular localization and trafficking of AnkG into the nucleus are dependent on binding to p32. Furthermore, we demonstrated that nuclear localization of AnkG but not binding to p32 is required for apoptosis inhibition. Thus, the antiapoptotic activity of AnkG is controlled by p32-mediated intracellular trafficking, which, in turn, seems to be regulated by host cell processes that sense stress.


Subject(s)
Apoptosis/drug effects , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Coxiella burnetii/metabolism , Animals , Bacterial Proteins/genetics , Cells, Cultured , Cricetinae , Escherichia coli/physiology , Gene Expression Regulation, Bacterial/physiology , Humans , Legionella pneumophila/physiology , Mice , Protein Transport
4.
J Immunol ; 180(11): 7546-52, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18490755

ABSTRACT

Recognition of LPS by TLR4 initiates inflammatory responses inducing potent antimicrobial immunity. However, uncontrolled inflammatory responses can be detrimental. To prevent the development of septic shock during an infection with Gram-negative bacteria, the immune system has developed mechanisms to neutralize LPS by specialized proteins. In this study, we report the recombinant expression and functional characterization of the mouse homolog of human bactericidal/permeability-increasing protein (BPI). Purified recombinant mouse BPI was able to neutralize LPS-mediated activation of macrophages and to block LPS-dependent maturation of dendritic cells. Recombinant mouse BPI neutralized the capacity of Gram-negative bacteria to activate immune cells, but did not influence the stimulatory properties of Gram-positive bacteria. Unlike human BPI, mouse BPI failed to kill or inhibit the growth of Pseudomonas aeruginosa. Together, these data demonstrate that murine BPI is a potent LPS-neutralizing protein that may limit innate immune responses during Gram-negative infections.


Subject(s)
Gram-Negative Bacteria/immunology , Lipopolysaccharides/immunology , Proteins/metabolism , Animals , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/metabolism , Blood Proteins/immunology , Blood Proteins/metabolism , Cell Line , Dendritic Cells/immunology , Escherichia coli/growth & development , Escherichia coli/immunology , Escherichia coli/pathogenicity , Gram-Negative Bacteria/metabolism , Humans , Macrophage Activation , Mice , Mice, Inbred C57BL , Proteins/immunology , Proteins/isolation & purification , Pseudomonas aeruginosa/growth & development , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
5.
J Immunol ; 176(1): 522-8, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16365446

ABSTRACT

Antimicrobial effector proteins are a key mechanism for the innate immune system to combat pathogens once they infect the host. We report the identification and cloning of the mouse homologue of human bactericidal permeability/increasing protein (BPI). Mouse BPI is constitutively expressed in lymphatic organs and tissues as well as in mouse testis. Upon stimulation with different TLR ligands, mouse BPI is strongly expressed in granulocytes and, surprisingly, in bone marrow-derived dendritic cells. Mouse BPI is most strongly induced by bacterial LPS through a signaling pathway that is completely dependent on TLR4-Toll/IL-1R domain-containing adaptor inducing IFN-beta. Functional studies revealed that mouse BPI does have the potential to neutralize LPS and inhibits bacterial growth. Mouse BPI is expressed in granulocytes and bone marrow-derived dendritic cells, and the transcriptional activation is controlled by TLRs.


Subject(s)
Blood Proteins/genetics , Blood Proteins/immunology , Endotoxins/metabolism , Interferon-beta/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , Receptors, Interleukin-1/metabolism , Toll-Like Receptors/metabolism , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Vesicular Transport/immunology , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides , Blood Proteins/metabolism , Blotting, Northern , Bone Marrow Cells/immunology , Cloning, Molecular , Dendritic Cells/immunology , Granulocytes/immunology , Granulocytes/metabolism , Humans , Interferon-beta/immunology , Lipopolysaccharides/immunology , Macrophages/immunology , Macrophages/microbiology , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Myeloid Differentiation Factor 88 , RNA, Messenger/analysis , Receptors, Interleukin-1/immunology , Reverse Transcriptase Polymerase Chain Reaction , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Sequence Homology, Amino Acid , Toll-Like Receptors/immunology , Transcription, Genetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...